1
|
Elimam H, Gauvin J, Huynh DN, Ménard L, Al-Hawat ML, Harb D, Lubell WD, Carpentier AC, Ong H, Marleau S. Targeting CD36 With EP 80317 Reduces Remote Inflammatory Response to Hind Limb Ischemia-Reperfusion in Mice. J Biochem Mol Toxicol 2024; 38:e70057. [PMID: 39552437 PMCID: PMC11582942 DOI: 10.1002/jbt.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
Reperfusion of ischemic skeletal muscle triggers oxidative stress and an immediate inflammatory reaction, leading to damage of distant organs such as the lungs. The inflammatory process implicates numerous mediators, including cytokines, chemokines, and arachidonic acid metabolites. In the orchestration of the inflammatory cascade, a critical role is played by the cluster of differentiation-36 receptor (CD36), a scavenger receptor class B protein (SR-B2) which is expressed on macrophages and functions as a Toll-like receptor coreceptor. A mouse model of hind limb ischemia-reperfusion has been used to investigate the interplay between CD36 signaling and remote inflammation: leukocyte recruitment, regulation of the nucleotide-binding domain leucin-rich repeat and pyrin-containing receptor 3 (NLRP3) inflammasome, and release of nuclear factor-kappa B (NF-ĸB) and arachidonic acid metabolites. Levels of reactive oxygen species, inflammatory mediators, and gene expression were measured in blood and lung tissue samples collected from anesthetized mice on which unilateral hind limb ischemia was induced by rubber band constriction for 30 min followed by reperfusion for 3 h. The CD36 modulator EP 80317, a member of the growth hormone releasing peptide 6 family, was employed as a pharmacological agent to mitigate distant lung injury following skeletal limb ischemia-reperfusion. Targeting CD36 on monocytes/macrophages, EP 80317 abated pro-inflammatory signaling and transcriptional activity encompassing lipid and cytokine mediators. Targeting CD36 was shown to offer promise for curtailing tissue injury following hind limb ischemia-reperfusion.
Collapse
Affiliation(s)
- Hanan Elimam
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Jade Gauvin
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - David N Huynh
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Liliane Ménard
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | | | - Diala Harb
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - André C Carpentier
- Department of Medicine, Division of Endocrinology, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
2
|
Toapanta FR, Hu J, Shirey KA, Bernal PJ, Levine MM, Darton TC, Waddington CS, Pollard AJ, Sztein MB. Changes in monocyte subsets in volunteers who received an oral wild-type Salmonella Typhi challenge and reached typhoid diagnosis criteria. Front Immunol 2024; 15:1454857. [PMID: 39263222 PMCID: PMC11388309 DOI: 10.3389/fimmu.2024.1454857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
An oral Controlled Human Infection Model (CHIM) with wild-type S. Typhi was re-established allowing us to explore the development of immunity. In this model, ~55% of volunteers who received the challenge reached typhoid diagnosis criteria (TD), while ~45% did not (NoTD). Intestinal macrophages are one of the first lines of defense against enteric pathogens. Most organs have self-renewing macrophages derived from tissue-resident progenitor cells seeded during the embryonic stage; however, the gut lacks these progenitors, and all intestinal macrophages are derived from circulating monocytes. After infecting gut-associated lymphoid tissues underlying microfold (M) cells, S. Typhi causes a primary bacteremia seeding organs of the reticuloendothelial system. Following days of incubation, a second bacteremia and clinical disease ensue. S. Typhi likely interacts with circulating monocytes or their progenitors in the bone marrow. We assessed changes in circulating monocytes after CHIM. The timepoints studied included 0 hours (pre-challenge) and days 1, 2, 4, 7, 9, 14, 21 and 28 after challenge. TD participants provided extra samples at the time of typhoid diagnosis, and 48-96 hours later (referred as ToD). We report changes in Classical Monocytes -CM-, Intermediate Monocytes -IM- and Non-classical Monocytes -NCM-. Changes in monocyte activation markers were identified only in TD participants and during ToD. CM and IM upregulated molecules related to interaction with bacterial antigens (TLR4, TLR5, CD36 and CD206). Of importance, CM and IM showed enhanced binding of S. Typhi. Upregulation of inflammatory molecules like TNF-α were detected, but mechanisms involved in limiting inflammation were also activated (CD163 and CD354 downregulation). CM upregulated molecules to interact/modulate cells of the adaptive immunity, including T cells (HLA-DR, CD274 and CD86) and B cells (CD257). Both CM and IM showed potential to migrate to the gut as integrin α4β7 was upregulated. Unsupervised analysis revealed 7 dynamic cell clusters. Five of these belonged to CM showing that this is the main population activated during ToD. Overall, we provide new insights into the changes that diverse circulating monocyte subsets undergo after typhoid diagnosis, which might be important to control this disease since these cells will ultimately become intestinal macrophages once they reach the gut.
Collapse
Affiliation(s)
- Franklin R Toapanta
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jingping Hu
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Paula J Bernal
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Myron M Levine
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Thomas C Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Claire S Waddington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Marcelo B Sztein
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Li X, Jiang S, Wang B, He S, Guo X, Lin J, Wei Y. Integrated multi-omics analysis and machine learning developed diagnostic markers and prognostic model based on Efferocytosis-associated signatures for septic cardiomyopathy. Clin Immunol 2024; 265:110301. [PMID: 38944364 DOI: 10.1016/j.clim.2024.110301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024]
Abstract
Septic cardiomyopathy (SCM) is characterized by an abnormal inflammatory response and increased mortality. The role of efferocytosis in SCM is not well understood. We used integrated multi-omics analysis to explore the clinical and genetic roles of efferocytosis in SCM. We identified six module genes (ATP11C, CD36, CEBPB, MAPK3, MAPKAPK2, PECAM1) strongly associated with SCM, leading to an accurate predictive model. Subgroups defined by EFFscore exhibited distinct clinical features and immune infiltration levels. Survival analysis showed that the C1 subtype with a lower EFFscore had better survival outcomes. scRNA-seq analysis of peripheral blood mononuclear cells (PBMCs) from sepsis patients identified four genes (CEBPB, CD36, PECAM1, MAPKAPK2) associated with high EFFscores, highlighting their role in SCM. Molecular docking confirmed interactions between diagnostic genes and tamibarotene. Experimental validation supported our computational results. In conclusion, our study identifies a novel efferocytosis-related SCM subtype and diagnostic biomarkers, offering new insights for clinical diagnosis and therapy.
Collapse
Affiliation(s)
- Xuelian Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijiu Jiang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, The First Affifiliated Hospital, Shihezi University, Shihezi, Xinjiang, China
| | - Boyuan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaolin He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jibin Lin
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Sekulovic O, Gallagher C, Lee J, Hao L, Zinonos S, Tan CY, Anderson A, Kanevsky I. Evidence of Reduced Virulence and Increased Colonization Among Pneumococcal Isolates of Serotype 3 Clade II Lineage in Mice. J Infect Dis 2024; 230:e182-e188. [PMID: 39052735 PMCID: PMC11272092 DOI: 10.1093/infdis/jiae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 01/30/2024] Open
Abstract
Recent phylogenetic profiling of pneumococcal serotype 3 (Pn3) isolates revealed a dynamic interplay among major lineages with the emergence and global spread of a variant termed clade II. The cause of Pn3 clade II dissemination along with epidemiological and clinical ramifications are currently unknown. Here, we sought to explore biological characteristics of dominant Pn3 clades in a mouse model of pneumococcal invasive disease and carriage. Carriage and virulence potential were strain dependent with marked differences among clades. We found that clinical isolates from Pn3 clade II are less virulent and less invasive in mice compared to clade I isolates. We also observed that clade II isolates are carried for longer and at higher bacterial densities in mice compared to clade I isolates. Taken together, our data suggest that the epidemiological success of Pn3 clade II could be related to alterations in the pathogen's ability to cause invasive disease and to establish a robust carriage episode.
Collapse
Affiliation(s)
- Ognjen Sekulovic
- Pfizer Inc, Bacterial Vaccines and Technology, Pearl River, New York
| | - Caitlyn Gallagher
- Pfizer Inc, Bacterial Vaccines and Technology, Pearl River, New York
| | - Jonathan Lee
- Pfizer Inc, Bacterial Vaccines and Technology, Pearl River, New York
| | - Li Hao
- Pfizer Inc, Bacterial Vaccines and Technology, Pearl River, New York
| | - Stavros Zinonos
- Pfizer Inc, Bacterial Vaccines and Technology, Pearl River, New York
| | - Charles Y Tan
- Pfizer Inc, Early Clinical Development, Collegeville, Pennsylvania
| | | | - Isis Kanevsky
- Pfizer Inc, Bacterial Vaccines and Technology, Pearl River, New York
| |
Collapse
|
5
|
Meiling L, Yiran C, Xiaoli S, Kaihui C, Toshihiko H, Kikuji I, Kazunori M, Hattori S, Fujisaki H, Liu W, Ikejima T. Gelatin but not type I collagen promotes bacteria phagocytosis in PMA-treated U937 human lymphoma cells. Connect Tissue Res 2024; 65:170-185. [PMID: 38526028 DOI: 10.1080/03008207.2024.2330693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/09/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE Besides comprising scaffolding, extracellular matrix components modulate many biological processes including inflammation and cell differentiation. We previously found precoating cell plates with extracellular matrix collagen I, or its denatured product gelatin, causes aggregation of macrophage-like human lymphoma U937 cells, which are induced to differentiation by phorbol myristate treatment. In the present study, we investigated the influence of gelatin or collagen I precoating on the bacteria phagocytosis in PMA-stimulated U937 cells. MATERIALS AND METHODS Colony forming units of phagocytosed bacteria, Giemsa-staining of cells with phagocytosed bacteria, confocal microscopic and flow cytometric analysis of cells with phagocytosed FITC-labeled bacteria and non-bioactive latex beats were conducted. RESULTS Gelatin precoating enhances the phagocytosis of both Gram-negative and positive bacteria, as shown by the increased colony forming units of bacteria phagocytosed by cells, and increased intracellular bacteria observed after Giemsa-staining. But collagen I has no marked influence. Confocal microscopy reveals that both live and dead FITC-bacteria were phagocytosed more in the cells with gelatin-coating but not collagen-coating. Of note, both gelatin and collagen I coating had no influence on the phagocytosis of non-bioactive latex beads. Since gelatin-coating increases autophagy but collagen I has no such impact, we are curious about the role of autophagy. Inhibiting autophagy reduced the phagocytosis of bacteria, in cells with gelatin-coating, while stimulating autophagy enhanced phagocytosis. CONCLUSION This study finds the bacteria-phagocytosis stimulatory effect of gelatin in PMA-treated U937 cells and reveals the positive regulatory role of autophagy, predicting the potential use of gelatin products in anti-bacterial therapy.
Collapse
Affiliation(s)
- Li Meiling
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Chen Yiran
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Sun Xiaoli
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Chen Kaihui
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Hayashi Toshihiko
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Itoh Kikuji
- Biochemical Center, Japan SLC Inc., Shizuoka, Japan
| | - Mizuno Kazunori
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Giriyappagoudar M, Vastrad B, Horakeri R, Vastrad C. Study on Potential Differentially Expressed Genes in Idiopathic Pulmonary Fibrosis by Bioinformatics and Next-Generation Sequencing Data Analysis. Biomedicines 2023; 11:3109. [PMID: 38137330 PMCID: PMC10740779 DOI: 10.3390/biomedicines11123109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 12/24/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with reduced quality of life and earlier mortality, but its pathogenesis and key genes are still unclear. In this investigation, bioinformatics was used to deeply analyze the pathogenesis of IPF and related key genes, so as to investigate the potential molecular pathogenesis of IPF and provide guidance for clinical treatment. Next-generation sequencing dataset GSE213001 was obtained from Gene Expression Omnibus (GEO), and the differentially expressed genes (DEGs) were identified between IPF and normal control group. The DEGs between IPF and normal control group were screened with the DESeq2 package of R language. The Gene Ontology (GO) and REACTOME pathway enrichment analyses of the DEGs were performed. Using the g:Profiler, the function and pathway enrichment analyses of DEGs were performed. Then, a protein-protein interaction (PPI) network was constructed via the Integrated Interactions Database (IID) database. Cytoscape with Network Analyzer was used to identify the hub genes. miRNet and NetworkAnalyst databaseswereused to construct the targeted microRNAs (miRNAs), transcription factors (TFs), and small drug molecules. Finally, receiver operating characteristic (ROC) curve analysis was used to validate the hub genes. A total of 958 DEGs were screened out in this study, including 479 up regulated genes and 479 down regulated genes. Most of the DEGs were significantly enriched in response to stimulus, GPCR ligand binding, microtubule-based process, and defective GALNT3 causes HFTC. In combination with the results of the PPI network, miRNA-hub gene regulatory network and TF-hub gene regulatory network, hub genes including LRRK2, BMI1, EBP, MNDA, KBTBD7, KRT15, OTX1, TEKT4, SPAG8, and EFHC2 were selected. Cyclothiazide and rotigotinethe are predicted small drug molecules for IPF treatment. Our findings will contribute to identification of potential biomarkers and novel strategies for the treatment of IPF, and provide a novel strategy for clinical therapy.
Collapse
Affiliation(s)
- Muttanagouda Giriyappagoudar
- Department of Radiation Oncology, Karnataka Institute of Medical Sciences (KIMS), Hubballi 580022, Karnataka, India;
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. Socitey’s College of Pharmacy, Gadag 582101, Karnataka, India;
| | - Rajeshwari Horakeri
- Department of Computer Science, Govt First Grade College, Hubballi 580032, Karnataka, India;
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India
| |
Collapse
|
7
|
Slimmen LJM, Giacalone VD, Schofield C, Horati H, Manaï BHAN, Estevão SC, Garratt LW, Peng L, Tirouvanziam R, Janssens HM, Unger WWJ. Airway macrophages display decreased expression of receptors mediating and regulating scavenging in early cystic fibrosis lung disease. Front Immunol 2023; 14:1202009. [PMID: 37457715 PMCID: PMC10338875 DOI: 10.3389/fimmu.2023.1202009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Background Cystic fibrosis (CF) airway disease is characterized by chronic inflammation, featuring neutrophil influx to the lumen. Airway macrophages (AMs) can promote both inflammation and resolution, and are thus critical to maintaining and restoring homeostasis. CF AM functions, specifically scavenging activity and resolution of inflammation, have been shown to be impaired, yet underlying processes remain unknown. We hypothesized that impaired CF AM function results from an altered expression of receptors that mediate or regulate scavenging, and set out to investigate changes in expression of these markers during the early stages of CF lung disease. Methods Bronchoalveolar lavage fluid (BALF) was collected from 50 children with CF aged 1, 3 or 5 years. BALF cells were analyzed using flow cytometry. Expression levels of surface markers on AMs were expressed as median fluorescence intensities (MFI) or percentage of AMs positive for these markers. The effect of age and neutrophilic inflammation, among other variables, on marker expression was assessed with a multivariate linear regression model. Results AM expression of scavenger receptor CD163 decreased with age (p = 0.016) and was negatively correlated with BALF %neutrophils (r = -0.34, p = 0.016). AM expression of immune checkpoint molecule SIRPα also decreased with age (p = 0.0006), but did not correlate with BALF %neutrophils. Percentage of AMs expressing lipid scavenger CD36 was low overall (mean 20.1% ± 16.5) and did not correlate with other factors. Conversely, expression of immune checkpoint PD-1 was observed on the majority of AMs (mean PD-1pos 72.9% ± 11.8), but it, too, was not affected by age or BALF %neutrophils. Compared to matched blood monocytes, AMs had a higher expression of CD16, CD91, and PD-1, and a lower expression of CD163, SIRPα and CD36. Conclusion In BALF of preschool children with CF, higher age and/or increased neutrophilic inflammation coincided with decreased expression of scavenger receptors on AMs. Expression of scavenging receptors and regulators showed a distinctly different pattern in AMs compared to blood monocytes. These findings suggest AM capacity to counter inflammation and promote homeostasis reduces during initiation of CF airway disease and highlight new avenues of investigation into impaired CF AM function.
Collapse
Affiliation(s)
- Lisa J. M. Slimmen
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
- Laboratory of Pediatrics, Infection and Immunity Group, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Vincent D. Giacalone
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Craig Schofield
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Hamed Horati
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Badies H. A. N. Manaï
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Silvia C. Estevão
- Laboratory of Pediatrics, Infection and Immunity Group, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Luke W. Garratt
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Limin Peng
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Hettie M. Janssens
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Wendy W. J. Unger
- Laboratory of Pediatrics, Infection and Immunity Group, Department of Pediatrics, Erasmus University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
8
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
9
|
Gawish R, Maier B, Obermayer G, Watzenboeck ML, Gorki AD, Quattrone F, Farhat A, Lakovits K, Hladik A, Korosec A, Alimohammadi A, Mesteri I, Oberndorfer F, Oakley F, Brain J, Boon L, Lang I, Binder CJ, Knapp S. A neutrophil-B-cell axis impacts tissue damage control in a mouse model of intraabdominal bacterial infection via Cxcr4. eLife 2022; 11:e78291. [PMID: 36178806 PMCID: PMC9525059 DOI: 10.7554/elife.78291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a life-threatening condition characterized by uncontrolled systemic inflammation and coagulation, leading to multiorgan failure. Therapeutic options to prevent sepsis-associated immunopathology remain scarce. Here, we established a mouse model of long-lasting disease tolerance during severe sepsis, manifested by diminished immunothrombosis and organ damage in spite of a high pathogen burden. We found that both neutrophils and B cells emerged as key regulators of tissue integrity. Enduring changes in the transcriptional profile of neutrophils include upregulated Cxcr4 expression in protected, tolerant hosts. Neutrophil Cxcr4 upregulation required the presence of B cells, suggesting that B cells promoted disease tolerance by improving tissue damage control via the suppression of neutrophils' tissue-damaging properties. Finally, therapeutic administration of a Cxcr4 agonist successfully promoted tissue damage control and prevented liver damage during sepsis. Our findings highlight the importance of a critical B-cell/neutrophil interaction during sepsis and establish neutrophil Cxcr4 activation as a potential means to promote disease tolerance during sepsis.
Collapse
Affiliation(s)
- Riem Gawish
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Barbara Maier
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Georg Obermayer
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - Martin L Watzenboeck
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Anna-Dorothea Gorki
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Federica Quattrone
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Asma Farhat
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Karin Lakovits
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
| | - Anastasiya Hladik
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
| | - Ana Korosec
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
| | - Arman Alimohammadi
- Department of Medicine II, Division of Cardiology, Medical University of ViennaViennaAustria
| | - Ildiko Mesteri
- Department of Pathology, Medical University ViennaViennaAustria
| | | | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle UniversityNewcastleUnited Kingdom
| | - John Brain
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle UniversityNewcastleUnited Kingdom
| | | | - Irene Lang
- Department of Medicine II, Division of Cardiology, Medical University of ViennaViennaAustria
| | - Christoph J Binder
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - Sylvia Knapp
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| |
Collapse
|
10
|
P. Tavares L, Brüggemann TR, M. Rezende R, G. Machado M, Cagnina RE, Shay AE, C. Garcia C, Nijmeh J, M. Teixeira M, Levy BD. Cysteinyl Maresins Reprogram Macrophages to Protect Mice from Streptococcus pneumoniae after Influenza A Virus Infection. mBio 2022; 13:e0126722. [PMID: 35913160 PMCID: PMC9426576 DOI: 10.1128/mbio.01267-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 12/03/2022] Open
Abstract
Influenza A virus (IAV) infections are a leading cause of mortality worldwide. Excess mortality during IAV epidemics and pandemics is attributable to secondary bacterial infections, particularly pneumonia caused by Streptococcus pneumoniae. Resident alveolar macrophages (rAMs) are early responders to respiratory infections that coordinate initial host defense responses. Maresin conjugates in tissue regeneration (MCTRs) are recently elucidated cysteinyl maresins that are produced by and act on macrophages. Roles for MCTRs in responses to respiratory infections remain to be determined. Here, IAV infection led to transient decreases in rAM numbers. Repopulated lung macrophages displayed transcriptional alterations 21 days post-IAV with prolonged susceptibility to secondary pneumococcal infection. Administration of a mix of MCTR1 to 3 or MCTR3 alone post-IAV decreased lung inflammation and bacterial load 48 and 72 h after secondary pneumococcal infection. MCTR-exposed rAMs had increased migration and phagocytosis of Streptococcus pneumoniae, reduced secretion of CXCL1, and a reversion toward baseline levels of several IAV-induced pneumonia susceptibility genes. Together, MCTRs counter regulated post-IAV changes in rAMs to promote a rapid return of bacteria host defense. IMPORTANCE Secondary bacterial pneumonia is a serious and common complication of IAV infection, leading to excess morbidity and mortality. New host-directed approaches are needed to complement antibiotics to better address this important global infectious disease. Here, we show that harnessing endogenous resolution mechanisms for inflammation by exogenous administration of a family of specialized proresolving mediators (i.e., cys-MCTRs) increased macrophage resilience mechanisms after IAV to protect against secondary infection from Streptococcus pneumoniae.
Collapse
Affiliation(s)
- Luciana P. Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thayse R. Brüggemann
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marina G. Machado
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - R. Elaine Cagnina
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ashley E. Shay
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cristiana C. Garcia
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julie Nijmeh
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauro M. Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruce D. Levy
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Li H, Fu Q, Philips K, Sun Y, Faurot KR, Gaylord SA, Mann JD. Leukocyte inflammatory phenotype and function in migraine patients compared with matched non-migraine volunteers: a pilot study. BMC Neurol 2022; 22:278. [PMID: 35896985 PMCID: PMC9327171 DOI: 10.1186/s12883-022-02781-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/30/2022] [Indexed: 11/12/2022] Open
Abstract
Background Migraine is a neurological condition characterized by chronic inflammation. However, not much is known about the potential role of peripheral blood immune cells in the pathophysiology of migraine. Methods We investigated the status of peripheral blood immune cells of 15 adults with frequent episodic or chronic migraine recruited chronologically from a randomized clinical trial (RCT) on Nutrition for Migraine (NCCIH 5R01AT007813-05) and 15 non-migraine, healthy volunteers (control) matched by age, gender, and Body Mass Index (BMI). Continuous variables were presented as means ± standard deviationas well as medians, and comparisons between patients and healthy volunteers were performed with non-parametric Wilcoxon signed rank tests. Statistical analysis was performed using Stata (StataCorp. 2019. Stata Statistical Software). Fluorescence-Activated Cell Sorting (FACS) data were processed using FlowJo software (Ashland, OR: Becton, Dickenson and Company; 2019). Results We observed that migraineurs had a significantly lower percentage of non-classical monocytes (CD14+CD16++) in blood circulation, compared to the control group. In addition, Migraineurs also showed a significantly lower percentage of blood CD3+CD4+ helper T cells and CD4+CD25+ regulatory T cells, compared to controls. Differences in leukocyte surface markers between chronic migraine patients and their matched controls were more prominent than those between episodic migraine patients and their matched controls. Conclusions Our results suggest that migraine is associated with dysregulated peripheral immune homeostasis and that inflammation and autoimmunity may play a role in its pathophysiology. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-022-02781-4.
Collapse
Affiliation(s)
- Hongtao Li
- Department of Science and Mathematics, Hulman Hall Room116, Saint Mary-of-the-Woods College, 1 St Mary of Woods Coll, IN, 47876, USA. .,Department of Physical Medicine and Rehabilitation Program On Integrative Medicine, University of North Carolina, Chapel Hill, NC, USA.
| | - Qiang Fu
- School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Kamaira Philips
- Division of Comprehensive Oral Health, Adams School of Dentistry, University of North Carolina, Chapel Hill, NC, USA
| | - Yufei Sun
- School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Keturah R Faurot
- Department of Physical Medicine and Rehabilitation Program On Integrative Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Susan A Gaylord
- Department of Physical Medicine and Rehabilitation Program On Integrative Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - John Douglas Mann
- Department of Physical Medicine and Rehabilitation Program On Integrative Medicine, University of North Carolina, Chapel Hill, NC, USA.,Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Shirato K, Sato S. Macrophage Meets the Circadian Clock: Implication of the Circadian Clock in the Role of Macrophages in Acute Lower Respiratory Tract Infection. Front Cell Infect Microbiol 2022; 12:826738. [PMID: 35281442 PMCID: PMC8904936 DOI: 10.3389/fcimb.2022.826738] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The circadian rhythm is a biological system that creates daily variations of physiology and behavior with a 24-h cycle, which is precisely controlled by the molecular circadian clock. The circadian clock dominates temporal activity of physiological homeostasis at the molecular level, including endocrine secretion, metabolic, immune response, coupled with extrinsic environmental cues (e.g., light/dark cycles) and behavioral cues (e.g., sleep/wake cycles and feeding/fasting cycles). The other side of the clock is that the misaligned circadian rhythm contributes to the onset of a variety of diseases, such as cancer, metabolic diseases, and cardiovascular diseases, the acceleration of aging, and the development of systemic inflammation. The role played by macrophages is a key mediator between circadian disruption and systemic inflammation. At the molecular level, macrophage functions are under the direct control of the circadian clock, and thus the circadian misalignment remodels the phenotype of macrophages toward a ‘killer’ mode. Remarkably, the inflammatory macrophages induce systemic and chronic inflammation, leading to the development of inflammatory diseases and the dampened immune defensive machinery against infectious diseases such as COVID-19. Here, we discuss how the circadian clock regulates macrophage immune functions and provide the potential risk of misaligned circadian rhythms against inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University School of Medicine, Mitaka, Japan
| | - Shogo Sato
- Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, TX, United States
- *Correspondence: Shogo Sato,
| |
Collapse
|
13
|
Peruń A, Gębicka M, Biedroń R, Skalska P, Józefowski S. The CD36 and SR-A/CD204 scavenger receptors fine-tune Staphylococcus aureus-stimulated cytokine production in mouse macrophages. Cell Immunol 2022; 372:104483. [DOI: 10.1016/j.cellimm.2022.104483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/20/2021] [Accepted: 01/11/2022] [Indexed: 11/03/2022]
|
14
|
Gautam I, Storad Z, Filipiak L, Huss C, Meikle CK, Worth RG, Wuescher LM. From Classical to Unconventional: The Immune Receptors Facilitating Platelet Responses to Infection and Inflammation. BIOLOGY 2020; 9:E343. [PMID: 33092021 PMCID: PMC7589078 DOI: 10.3390/biology9100343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/06/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
Platelets have long been recognized for their role in maintaining the balance between hemostasis and thrombosis. While their contributions to blood clotting have been well established, it has been increasingly evident that their roles extend to both innate and adaptive immune functions during infection and inflammation. In this comprehensive review, we describe the various ways in which platelets interact with different microbes and elicit immune responses either directly, or through modulation of leukocyte behaviors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leah M. Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (I.G.); (Z.S.); (L.F.); (C.H.); (C.K.M.); (R.G.W.)
| |
Collapse
|
15
|
Serpa GL, Renton ND, Lee N, Crane MJ, Jamieson AM. Electronic Nicotine Delivery System Aerosol-induced Cell Death and Dysfunction in Macrophages and Lung Epithelial Cells. Am J Respir Cell Mol Biol 2020; 63:306-316. [PMID: 32469619 DOI: 10.1165/rcmb.2019-0200oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Electronic nicotine delivery system (ENDS) use is outpacing our understanding of its potential harmful effects. Homeostasis of the lung is maintained through proper balance of cell death, efferocytic clearance, and phagocytosis of pathogens. To investigate whether ENDS use has the potential to alter this balance, we developed physiologically relevant ENDS exposure paradigms for lung epithelial cells and primary macrophages. In our studies, cells were exposed directly to aerosol made from carefully controlled components with and without nicotine. We found that ENDS aerosol exposure led to apoptosis, secondary necrosis, and necrosis in lung epithelial cell models. In contrast, macrophages died mostly by apoptosis and inflammatory caspase-mediated cell death when exposed to ENDS aerosol. The clearance of dead cells and pathogens by efferocytosis and phagocytosis, respectively, is an important process in maintaining a healthy lung. To investigate the impact of ENDS aerosol on macrophage function independent of general toxicity, we used an exposure time that did not induce cell death in primary macrophages. Exposure to ENDS aerosol containing nicotine inhibited nearly all phagocytic and greatly reduced the efferocytic abilities of primary macrophages. When challenged with a bacterial pathogen, there was decreased bacterial clearance. The presence of nicotine in the ENDS aerosol increased its toxicity and functional impact; however, nicotine exposure alone did not have any deleterious effects. These data demonstrate that ENDS aerosol exposure could lead to increased epithelial cell and macrophage death in the lung and impair important macrophage functions that are essential for maintenance of lung function.
Collapse
Affiliation(s)
- Gregory L Serpa
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Nicholas D Renton
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Nari Lee
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Meredith J Crane
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Amanda M Jamieson
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| |
Collapse
|
16
|
Banesh S, Trivedi V. Therapeutic Potentials of Scavenger Receptor CD36 Mediated Innate Immune Responses Against Infectious and Non-Infectious Diseases. Curr Drug Discov Technol 2020; 17:299-317. [PMID: 31376823 DOI: 10.2174/1570163816666190802153319] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/18/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
CD36 is a multifunctional glycoprotein, expressed in different types of cells and known to play a significant role in the pathophysiology of the host. The structural studies revealed that the scavenger receptor consists of short cytosolic domains, two transmembrane domains, and a large ectodomain. The ectodomain serves as a receptor for a diverse number of endogenous and exogenous ligands. The CD36-specific ligands are involved in regulating the immune response during infectious and non-infectious diseases in the host. The role of CD36 in regulating the innate immune response during Pneumonia, Tuberculosis, Malaria, Leishmaniasis, HIV, and Sepsis in a ligand- mediated fashion. Apart from infectious diseases, it is also considered to be involved in metabolic disorders such as Atherosclerosis, Alzheimer's, cancer, and Diabetes. The ligand binding to scavenger receptor modulates the CD36 down-stream innate immune response, and it can be exploited to design suitable immuno-modulators. Hence, the current review focused on the role of the CD36 in innate immune response and therapeutic potentials of novel heterocyclic compounds as CD36 ligands during infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Sooram Banesh
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| |
Collapse
|
17
|
Che Z, Shao Y, Zhang W, Zhao X, Guo M, Li C. Cloning and functional analysis of scavenger receptor B gene from the sea cucumber Apostichopus japonicus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 99:103404. [PMID: 31152761 DOI: 10.1016/j.dci.2019.103404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 06/09/2023]
Abstract
Scavenger receptor (SR) class B (SR-B) is a transmembrane protein that belongs to the SR family with a wide range of functions in innate immunity. Here, an SR-B homologue, designated as AjSR-B, was cloned from the sea cucumber Apostichopus japonicus. AjSR-B comprised 2519 nucleotides with a 5'-untranslated region (UTR) of 153 bp, an open reading frame of 1581 bp encoding a 526 amino acid protein, and a 3'-UTR of 785 bp. SMART analysis indicated that AjSR-B has two transmembrane regions and a cluster determinant 36 domain. Multiple alignments and phylogenetic analysis supported that AjSR-B is a novel member of the SR-B protein family. Moreover, AjSR-B was constitutively expressed in all detected tissues, with the highest levels recorded in the intestine. Both were significantly induced in coelomocytes and the intestine after Vibrio splendidus challenge. Functionally, the recombinant rAjSR-B that corresponds to a large extracellular loop can bind pathogen-associated molecular patterns (PAMPs), including lipopolysaccharide (LPS), peptidoglycan, and mannan, with a high binding affinity to LPS. Bacterial agglutination assay showed that rAjSR-B can agglutinate the four tested bacteria (Gram-negative and Gram-positive bacteria) with calcium dependence. However, the agglutination ability for Gram-negative bacteria completely disappeared in the presence of PAMPs but a weak ability to bind Gram-positive bacteria (Micrococcus luteus) was still exhibited, suggesting there might exist a competition between Gram-positive bacteria and PAMPs under same condition. Our current study indicated that AjSR-B is a PAMP that plays important roles in the innate immune process of sea cucumbers.
Collapse
Affiliation(s)
- Zhongjie Che
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Ming Guo
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
18
|
Olotu C, Lehmensiek F, Koch B, Kiefmann M, Riegel AK, Hammerschmidt S, Kiefmann R. Streptococcus pneumoniae inhibits purinergic signaling and promotes purinergic receptor P2Y 2 internalization in alveolar epithelial cells. J Biol Chem 2019; 294:12795-12806. [PMID: 31289122 DOI: 10.1074/jbc.ra118.007236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Bacterial pneumonia is a global health challenge that causes up to 2 million deaths each year. Purinergic signaling plays a pivotal role in healthy alveolar epithelium. Here, we used fluorophore-based analysis and live-cell calcium imaging to address the question of whether the bacterial pathogen Streptococcus pneumoniae directly interferes with purinergic signaling in alveolar epithelial cells. Disturbed purinergic signaling might result in pathophysiologic changes like edema formation and atelectasis, which are commonly seen in bacterial pneumonia. Purine receptors are mainly activated by ATP, mediating a cytosolic calcium response. We found that this purinergic receptor P2Y2-mediated response is suppressed in the presence of S. pneumoniae in A549 and isolated primary alveolar cells in a temperature-dependent manner. Downstream inositol 3-phosphate (IP3) signaling appeared to be unaffected, as calcium signaling via protease-activated receptor 2 remained unaltered. S. pneumoniae-induced suppression of the P2Y2-mediated calcium response depended on the P2Y2 phosphorylation sites Ser-243, Thr-344, and Ser-356, which are involved in receptor desensitization and internalization. Spinning-disk live-cell imaging revealed that S. pneumoniae induces P2Y2 translocation into the cytosol. In conclusion, our results show that S. pneumoniae directly inhibits purinergic signaling by inducing P2Y2 phosphorylation and internalization, resulting in the suppression of the calcium response of alveolar epithelial cells to ATP, thereby affecting cellular integrity and function.
Collapse
Affiliation(s)
- Cynthia Olotu
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22051 Hamburg, Germany
| | - Felix Lehmensiek
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22051 Hamburg, Germany
| | - Bastian Koch
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22051 Hamburg, Germany
| | - Martina Kiefmann
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22051 Hamburg, Germany
| | - Ann-Kathrin Riegel
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22051 Hamburg, Germany
| | - Sven Hammerschmidt
- Institute of Genetics and Functional Genomics, Department of Molecular Genetics and Infection Biology, University of Greifswald, Felix-Hausdorff-Strasse 8, 17489 Greifswald, Germany
| | - Rainer Kiefmann
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22051 Hamburg, Germany
| |
Collapse
|
19
|
Pombinho R, Sousa S, Cabanes D. Scavenger Receptors: Promiscuous Players during Microbial Pathogenesis. Crit Rev Microbiol 2018; 44:685-700. [PMID: 30318962 DOI: 10.1080/1040841x.2018.1493716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Innate immunity is the most broadly effective host defense, being essential to clear the majority of microbial infections. Scavenger Receptors comprise a family of sensors expressed in a multitude of host cells, whose dual role during microbial pathogenesis gained importance over recent years. SRs regulate the recruitment of immune cells and control both host inflammatory response and bacterial load. In turn, pathogens have evolved different strategies to overcome immune response, avoid recognition by SRs and exploit them to favor infection. Here, we discuss the most relevant findings regarding the interplay between SRs and pathogens, discussing how these multifunctional proteins recognize a panoply of ligands and act as bacterial phagocytic receptors.
Collapse
Affiliation(s)
- Rita Pombinho
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| | - Sandra Sousa
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| | - Didier Cabanes
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| |
Collapse
|
20
|
Yang H, Li Q, Wang C, Wang J, Lv J, Wang L, Zhang ZS, Yao Z, Wang Q. Cytotoxic Necrotizing Factor 1 Downregulates CD36 Transcription in Macrophages to Induce Inflammation During Acute Urinary Tract Infections. Front Immunol 2018; 9:1987. [PMID: 30233583 PMCID: PMC6128224 DOI: 10.3389/fimmu.2018.01987] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 08/13/2018] [Indexed: 11/13/2022] Open
Abstract
Urinary tract infections (UTIs) caused by uropathogenic Escherichia coli (UPEC) induce cystitis, pyelonephritis, and can cause kidney scarring and failure if inflammation is not under control. The detailed effects of cytotoxic necrotizing factor 1 (CNF1), the key UPEC toxin, on the pathogenicity of UPEC remain unclear. CD36 is an important scavenger receptor, responsible for pathogen and apoptotic cell clearance, and plays an essential role in host immune defense and homeostasis. Regulation of CD36 by bacterial toxins has not been reported. In this study, using a pyelonephritis mouse model, CNF1 was observed to contribute to increasing neutrophils and bacterial titers in infected bladder and kidney tissues, resulting in severe inflammation and tissue damage. CD36 expression in macrophages was found to be decreased by CNF1 in vitro and in vivo. We demonstrated that CNF1 attenuated CD36 transcription by decreasing expressions of its upstream transcription factors LXRβ and C/EBPα and their recruitment to the CD36 promotor. In addition, Cdc42 was found to be involved in CNF1-mediated downregulation of LXRβ. Our study investigated the pathogenesis of cnf1-carrying UPEC, which affected host innate immune defenses and homeostasis through regulation of CD36 in macrophages during acute UTIs.
Collapse
Affiliation(s)
- Huan Yang
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qianqian Li
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Changying Wang
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jingyu Wang
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junqiang Lv
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lei Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Collaborative Innovation Center for Biotherapy, College of Pharmacy, Nankai University, Tianjin, China
| | - Zhi-Song Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Collaborative Innovation Center for Biotherapy, College of Pharmacy, Nankai University, Tianjin, China
| | - Zhi Yao
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Quan Wang
- Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
21
|
Kong T, Gong Y, Liu Y, Wen X, Tran NT, Aweya JJ, Zhang Y, Ma H, Zheng H, Li S. Scavenger receptor B promotes bacteria clearance by enhancing phagocytosis and attenuates white spot syndrome virus proliferation in Scylla paramamosian. FISH & SHELLFISH IMMUNOLOGY 2018; 78:79-90. [PMID: 29679762 DOI: 10.1016/j.fsi.2018.04.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 06/08/2023]
Abstract
Phagocytosis and apoptosis are key cellular innate immune responses against bacteria and virus in invertebrates. Class B scavenger receptors (SRBs), which contain a CD36 domain, are critical pattern recognition receptors (PRRs) of phagocytosis for bacteria and apoptotic cells. In the present study, we identified a member of SRB subfamily in mud crab Scylla paramamosain, named Sp-SRB. The full-length cDNA of Sp-SRB is 2593 bp with a 1629 bp open reading frame (ORF) encoding a putative protein of 542 amino acids, and predicted to contain a CD36 domain with two transmembrane regions at the C- and N-terminals. Real-time qPCR analysis revealed that Sp-SRB was widely expressed in all tissues tested, and the expression of Sp-SRB was up-regulated upon challenge with Vibrio parahaemolyticus, white spot syndrome virus (WSSV), lipopolysaccharides (LPS) and polyinosinic polycytidylic acid (PolyI:C). Moreover, in vitro experiments indicated that recombinant Sp-SRB protein (rSp-SRB) could bind to fungi, Gram-positive and Gram-negative bacteria. RNA interference of Sp-SRB resulted in significant reduction in the expression level of phagocytosis related genes, antimicrobial peptides (AMPs) and Toll-like receptors (TLRs), which consequently led to impairment in both bacterial clearance and the phagocytotic activity of hemocytes. In addition, we found that Sp-SRB had the ability to attenuate the replication of WSSV proliferation in mud crab S. paramamosain. Collectively, this study has shown that Sp-SRB contributed to bacteria clearance by enhancing phagocytosis and up-regulating the expression of AMPs possibly in a TRLs (SpToll 1 and SpToll 2)-dependent manner. Besides, Sp-SRB inhibited the replication of WSSV in S. paramamosian probably through enhancement of hemocytes phagocytosis of apoptotic cells.
Collapse
Affiliation(s)
- Tongtong Kong
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Yi Gong
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Yan Liu
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Xiaobo Wen
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Ngoc Tuan Tran
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Jude Juventus Aweya
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Hongyu Ma
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Huaiping Zheng
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China
| | - Shengkang Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Marine Biology Institute, Shantou University, Shantou, 515063, China.
| |
Collapse
|
22
|
Ye W, Zhang J, Shu Z, Yin Y, Zhang X, Wu K. Pneumococcal LytR Protein Is Required for the Surface Attachment of Both Capsular Polysaccharide and Teichoic Acids: Essential for Pneumococcal Virulence. Front Microbiol 2018; 9:1199. [PMID: 29951042 PMCID: PMC6008509 DOI: 10.3389/fmicb.2018.01199] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/16/2018] [Indexed: 11/13/2022] Open
Abstract
The LytR-Cps-Psr family proteins are commonly present in Gram-positive bacteria, which have been shown to implicate in anchoring cell wall-related glycopolymers to the peptidoglycan. Here, we report the cellular function of SPD_1741 (LytR) in Streptococcus pneumoniae and its role in virulence of pneumococci. Pneumococcal ΔlytR mutants have been successfully constructed by replacing the lytR gene with erm cassette. The role of LytR in pneumococcal growth was determined by growth experiments, and surface accessibility of the LytR protein was analyzed using flow cytometry. Transmission electron microscopy (TEM) and immunoblotting were used to reveal the changes in capsular polysaccharide (CPS). Dot blot and ELISA were used to quantify the amount of teichoic acids (TAs). The contribution of LytR on bacterial virulence was assessed using in vitro phagocytosis assays and infection experiments. Compared to the wild-type strain, the ΔlytR mutant showed a defect in growth which merely grew to a maximal OD620 of 0.2 in the liquid medium. The growth of the ΔlytR mutant could be restored by addition of recombinant ΔTM-LytR protein in culture medium in a dose-dependent manner. TEM results showed that the D39ΔlytR mutant was impaired in the surface attachment of CPS. Deletion of lytR gene also impaired the retention of TAs on the surface of pneumococci. The reduction of CPS and TAs on the pneumocccal cells were confirmed using Dot blot and ELISA assays. Compared to wild-type D39, the ΔlytR mutant was more susceptible to the phagocytosis. Animal studies showed that the ability to colonize the nasophaynx and virulence of pneumococci were affected by impairment of the lytR gene. Collectively, these results suggest that pneumococcal LytR is involved in anchoring both the CPS and TAs to cell wall, which is important for virulence of pneumococci.
Collapse
Affiliation(s)
- Weijie Ye
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jinghui Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Zhaoche Shu
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Kaifeng Wu
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China.,Department of Laboratory Medicine, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
23
|
Olatunde AC, Abell LP, Landuyt AE, Hiltbold Schwartz E. Development of endocytosis, degradative activity, and antigen processing capacity during GM-CSF driven differentiation of murine bone marrow. PLoS One 2018; 13:e0196591. [PMID: 29746488 PMCID: PMC5944997 DOI: 10.1371/journal.pone.0196591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 04/16/2018] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DC) are sentinels of the immune system, alerting and enlisting T cells to clear pathogenic threats. As such, numerous studies have demonstrated their effective uptake and proteolytic activities coupled with antigen processing and presentation functions. Yet, less is known about how these cellular mechanisms change and develop as myeloid cells progress from progenitor cells to more differentiated cell types such as DC. Thus, our study comparatively examined these functions at different stages of myeloid cell development driven by the GM-CSF. To measure these activities at different stages of development, GM-CSF driven bone marrow cells were sorted based on expression of Ly6C, CD115, and CD11c. This strategy enables isolation of cells representing five distinct myeloid cell types: Common Myeloid Progenitor (CMP), Granulocyte/Macrophage Progenitor (GMP), monocytes, monocyte-derived Macrophage/monocyte-derived Dendritic cell Precursors (moMac/moDP), and monocyte-derived DC (moDC). We observed significant differences in the uptake capacity, proteolysis, and antigen processing and presentation functions between these myeloid cell populations. CMP showed minimal uptake capacity with no detectable antigen processing and presenting function. The GMP population showed higher uptake capacity, modest proteolytic activity, and little T cell stimulatory function. In the monocyte population, the uptake capacity reached its peak, yet this cell type had minimal antigen processing and presentation function. Finally, moMac/moDP and moDC had a modestly decreased uptake capacity, high degradative capacity and strong antigen processing and presentation functions. These insights into when antigen processing and presentation function develop in myeloid cells during GM-CSF driven differentiation are crucial to the development of vaccines, allowing targeting of the most qualified cells as an ideal vaccine vehicles.
Collapse
Affiliation(s)
| | - Laura P. Abell
- Department of Biological Sciences, Auburn University, Auburn, Alabama
| | - Ashley E. Landuyt
- Department of Biological Sciences, Auburn University, Auburn, Alabama
| | | |
Collapse
|
24
|
Abstract
The hydrophobicity of vitamin E poses transport and metabolic challenges to regulate its bioavailability and to prevent its accumulation in lipid-rich tissues such as adipose tissue, brain, and liver. Water-soluble precursors of vitamin E (α-tocopherol, αT), such as its esters with acetate (αTA), succinate (αTS), or phosphate (αTP), have increased solubility in water and stability against reaction with free radicals, but they are rapidly converted during their uptake into the lipid-soluble vitamin E. Therefore, the bioavailability of these precursors as intact molecules is low; nevertheless, at least for αTS and αTP, the recent research has revealed unique regulatory effects on signal transduction and gene expression and the modulation of cellular events ranging from proliferation, survival/apoptosis, lipid uptake and metabolism, phagocytosis, long term potentiation, cell migration, telomere maintenance, and angiogenesis. Moreover, water-soluble derivatives of vitamin E including some based on αTP are increasingly used as components of nanocarriers for enhanced and targeted delivery of drugs and other molecules (vitamins, including αT and αTP itself, vitamin D3, carnosine, caffeine, docosahexaenoic acid (DHA), insulin) and cofactors such as coenzyme Q10. In this review, the chemical characteristics, transport, metabolic pathways, and molecular mechanisms of action of αTP in cells and tissues are summarized and put into perspective with its possible role in the prevention of a number of diseases.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Miller School of Medicine, University of Miami, Miami, FL, United States.
| |
Collapse
|
25
|
Sun HL, Wu YR, Song FF, Gan J, Huang LY, Zhang L, Huang C. Role of PCSK9 in the Development of Mouse Periodontitis Before and After Treatment: A Double-Edged Sword. J Infect Dis 2017; 217:667-680. [DOI: 10.1093/infdis/jix574] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 12/01/2017] [Indexed: 01/30/2023] Open
Affiliation(s)
- Hua Ling Sun
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| | - Yan Ru Wu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| | - Fang Fang Song
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| | - Jing Gan
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| | - Li Yuan Huang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| | - Lu Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| | - Cui Huang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Hubei, People’s Republic of China
| |
Collapse
|
26
|
Wu YM, Yang L, Li XJ, Li L, Wang Q, Li WW. A class B scavenger receptor from Eriocheir sinensis (EsSR-B1) restricts bacteria proliferation by promoting phagocytosis. FISH & SHELLFISH IMMUNOLOGY 2017; 70:426-436. [PMID: 28916359 DOI: 10.1016/j.fsi.2017.09.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/04/2017] [Accepted: 09/09/2017] [Indexed: 06/07/2023]
Abstract
Scavenger receptors (SRs) are important pattern recognition receptors (PRRs), which play significant roles in host defense against pathogens by identifying pathogen-associated molecular patterns (PAMPs). In this study, we report the cloning and characterization of a SR from Eriocheir sinensis (EsSR-B1) which is a 500 amino acid protein encoded by a gene comprised of 2726 nucleotides with a 1503 bp open reading frame. The domains of EsSR-B1 were found to be evolutionarily conserved. EsSR-B1 was widely detected in different tissues of E. sinensis and significantly up-regulated in hemocytes after stimulation by Staphyloccocus aureus or Vibrio parahaemolyticus. Recombinant EsSR-B1 protein could bind to bacteria and promote phagocytosis upon bacterial stimulation. Moreover, antimicrobial peptide expression was reduced in EsSR-B1-silenced hemocytes after challenge by S. aureus or V. parahaemolyticus. Thus, EsSR-B1 has a critical role in the binding of bacteria and subsequent promotion of hemocyte phagocytosis.
Collapse
Affiliation(s)
- Yao-Meng Wu
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Lei Yang
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Xue-Jie Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Lu Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China.
| | - Wei-Wei Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China.
| |
Collapse
|
27
|
Yang MC, Yang HT, Li J, Sun JJ, Bi WJ, Niu GJ, Zhang Q, Shi XZ, Zhao XF, Wang JX. Scavenger receptor C promotes bacterial clearance in kuruma shrimp Marsupenaeus japonicus by enhancing hemocyte phagocytosis and AMP expression. FISH & SHELLFISH IMMUNOLOGY 2017; 67:254-262. [PMID: 28602682 DOI: 10.1016/j.fsi.2017.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/23/2017] [Accepted: 06/03/2017] [Indexed: 06/07/2023]
Abstract
Scavenger receptors (SRs) comprise a large family of structurally diverse glycoproteins located on the cell membrane and function as pattern-recognition receptors (PRRs) participating in innate immunity in different species. Class C scavenger receptor (SRC) has been only identified in invertebrates and its biological functions still need to be researched. In this study, we characterized the anti-bacterial function of a SRC from kuruma shrimp Marsupenaeus japonicus (MjSRC). The mRNA level of MjSRC was up-regulated significantly in hemocytes of kuruma shrimp challenged by Vibrio anguillarum or Staphylococcus aureus. The recombinant extracellular domains (MAM and CCP domains) of MjSRC have the ability of binding different bacteria and glycans in vitro. After knockdown of MjSRC, the bacterial clearance ability and phagocytic rate of hemocyte decreased significantly in vivo. Meanwhile, overexpression of MjSRC in shrimp enhanced the clearance ability and phagocytic rate of hemocytes. Further study found that MjSRC could regulate the expression of several antimicrobial peptides (AMPs). All these results indicate that MjSRC plays important roles in antibacterial immunity in kuruma shrimp by enhancing hemocyte phagocytosis and AMP expression.
Collapse
Affiliation(s)
- Ming-Chong Yang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Hui-Ting Yang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Jing Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Jie-Jie Sun
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Wen-Jie Bi
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Guo-Juan Niu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Qiang Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Xiu-Zhen Shi
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China.
| |
Collapse
|
28
|
Patel PS, Kearney JF. CD36 and Platelet-Activating Factor Receptor Promote House Dust Mite Allergy Development. THE JOURNAL OF IMMUNOLOGY 2017; 199:1184-1195. [PMID: 28667161 DOI: 10.4049/jimmunol.1700034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/07/2017] [Indexed: 12/21/2022]
Abstract
Over 89% of asthmatic children in underdeveloped countries demonstrate sensitivity to house dust mites (HDMs). The allergic response to HDMs is partially mediated by epithelial cell-derived cytokines that activate group 2 innate lymphoid cells, induce migration and activation of dendritic cells, and promote effector differentiation of HDM-specific TH2 cells. However, the contribution of innate receptor engagement on epithelial or dendritic cells by HDMs that ultimately mediates said innate and adaptive allergic responses is poorly understood. We and other investigators have demonstrated that HDMs express phosphorylcholine (PC) moieties. The major PC receptors involved in immune responses include CD36 and platelet-activating factor receptor (PAFR). Because CD36 and PAFR are expressed by epithelial cells and dendritic cells, and expression of these receptors is higher in human asthmatics, we determined whether engagement of CD36 or PAFR on epithelial or dendritic cells contributes to HDM allergy development. Testing bone marrow chimeric mice revealed that CD36 engagement on radioresistant cells and PAFR engagement on radioresistant and radiosensitive cells in the lung promote allergic responses to HDMs. Additionally, passive anti-PC IgM Abs administered intratracheally with HDMs decreased allergen uptake by epithelial cells and APCs in the lungs of C57BL/6 mice but not CD36-/- or PAFR-/- mice. These results show that CD36 and PAFR are important mediators of HDM allergy development and that inhibiting HDM engagement with PC receptors in the lung protects against allergic airway disease.
Collapse
Affiliation(s)
- Preeyam S Patel
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - John F Kearney
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
29
|
Zingg JM, Azzi A, Meydani M. α-Tocopheryl Phosphate Induces VEGF Expression via CD36/PI3Kγ in THP-1 Monocytes. J Cell Biochem 2017; 118:1855-1867. [PMID: 28059487 DOI: 10.1002/jcb.25871] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/05/2017] [Indexed: 12/20/2022]
Abstract
The CD36 scavenger receptor binds several ligands and mediates ligand uptake and ligand-dependent signal transduction and gene expression, events that may involve CD36 internalization. Here we show that CD36 internalization in THP-1 monocytes is triggered by α-tocopherol (αT) and more strongly by α-tocopheryl phosphate (αTP) and EPC-K1, a phosphate diester of αTP and L-ascorbic acid. αTP-triggered CD36 internalization is prevented by the specific covalent inhibitor of selective lipid transport by CD36, sulfo-N-succinimidyl oleate (SSO). Moreover, SSO inhibited the CD36-mediated uptake of 14C-labelled αTP suggesting that αTP binding and internalization of CD36 is involved in cellular αTP uptake, whereas the uptake of αT was less affected. Similar to that, inhibition of selective lipid transport of the SR-BI scavenger receptor resulted mainly in reduction of αTP and not αT uptake. In contrast, uptake of αT was mainly inhibited by Dynasore, an inhibitor of clathrin-mediated endocytosis, suggesting that the differential regulatory effects of αTP and αT on signaling may be influenced by their different routes of uptake. Interestingly, αTP and EPC-K1 also reduced the neutral lipid content of THP-1 cells and the phagocytosis of fluorescent Staphylococcus aureus bioparticles. Moreover, induction of the vascular endothelial growth factor (VEGF) promoter activity by αTP occurred via CD36/PI3Kγ/Akt, as it could be inhibited by specific inhibitors of this pathway (SSO, Wortmannin, AS-605240). These results suggest that αTP activates PI3Kγ/Akt signaling leading to VEGF expression in monocytes after binding to and/or transport by CD36, a receptor known to modulate angiogenesis in response to amyloid beta, oxLDL, and thrombospondin. J. Cell. Biochem. 118: 1855-1867, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Vascular Biology Laboratory, JM USDA-Human Nutr. Res. Ctr. on Aging, Tufts University, Boston, Massachusetts 02111
| | - Angelo Azzi
- Vascular Biology Laboratory, JM USDA-Human Nutr. Res. Ctr. on Aging, Tufts University, Boston, Massachusetts 02111
| | - Mohsen Meydani
- Vascular Biology Laboratory, JM USDA-Human Nutr. Res. Ctr. on Aging, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
30
|
Cao D, Luo J, Zang W, Chen D, Xu H, Shi H, Jing X. Gamma-Linolenic Acid Suppresses NF-κΒ Signaling via CD36 in the Lipopolysaccharide-Induced Inflammatory Response in Primary Goat Mammary Gland Epithelial Cells. Inflammation 2017; 39:1225-37. [PMID: 27121266 DOI: 10.1007/s10753-016-0358-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gamma-linolenic acid (GLA) and linoleic acid (LA), which are both n-6 unsaturated fatty acids, play vital roles in lipopolysaccharide (LPS)-induced inflammation. The multi-functional protein scavenger receptor CD36 has also been shown to participate in inflammation. However, the molecular mechanisms underlying the interactions between CD36 and GLA or LA in LPS-induced inflammation remain unclear. We used small interfering RNA and adenoviral systems to manipulate CD36 expression in primary goat mammary gland epithelial cells (pGMECs), and the results showed that nuclear factor kappa B (NF-κB) levels were significantly decreased by CD36 receptor signaling following treatment with GLA but not LA. GLA inhibited NF-κB activation in LPS-induced pGMECs. However, silencing CD36 or deleting its fatty acid-binding domain blocked the anti-inflammatory effects of GLA, resulting in an increase in NF-κB activation and disrupting its localization during LPS-induced inflammation. The activity of the cytokines IL-1β, IL-6, and TNF-α, which act downstream of NF-κB, was also modulated when CD34 expression was manipulated by the addition of GLA in LPS-induced pGMECs. Our data suggest that GLA, but not LA, may interact with the CD36 fatty acid-binding domain to regulate the activation and localization of NF-κB in LPS-induced pGMECs.
Collapse
Affiliation(s)
- Duoyao Cao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - WenJuan Zang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Dekun Chen
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Huifen Xu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Huaiping Shi
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaoqi Jing
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| |
Collapse
|
31
|
Abstract
The lung is constantly exposed to airborne infectious agents due to the large surface area of approximately 100 m2. Therefore pneumonia is one of the most common lung diseases. Understanding infection requires understanding the routes of infections, the way invading organisms infect epithelial cells, as well as defense mechanisms of the lung tissue acquired during evolution. Different variants of infectious and non-infectious pneumonias are discussed; special types of pneumonias such as granulomatous and fibrosing pneumonias are presented under separate sections. Causing organisms and other causes of pneumonias are included, and their mode of action is included as far as understood.
Collapse
|
32
|
Peruń A, Biedroń R, Konopiński MK, Białecka A, Marcinkiewicz J, Józefowski S. Phagocytosis of live versus killed or fluorescently labeled bacteria by macrophages differ in both magnitude and receptor specificity. Immunol Cell Biol 2016; 95:424-435. [PMID: 27826145 DOI: 10.1038/icb.2016.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 01/23/2023]
Abstract
Scavenger receptor (SR)-mediated opsonin-independent phagocytosis of bacteria by macrophages has been suggested to represent an important, early mechanism of anti-bacterial host defense. However, although the ability to bind bacteria has been demonstrated to be a shared feature of all types of SRs, in many cases the evidence is limited to the demonstration of increased binding of killed, fluorescently labeled bacteria to non-phagocytic cells transfected with these receptors. We sought to verify the ability of SRs to mediate non-opsonic phagocytosis of live Escherichia coli (Ec) and Staphylococcus aureus (Sa), model species of Gram-negative and -positive bacteria, respectively, and to assess the relative contributions of different SRs expressed on murine macrophages in this process. We found that the class A SR SR-A/CD204 was the major receptor mediating phagocytosis of fluorescently labeled Sa, whereas different SRs had highly redundant roles in the phagocytosis of live Sa. Conversely, different SRs contributed to the phagocytosis of fluorescently labeled Ec. In comparison, phagocytosis of live Ec was of much lower magnitude and was selectively mediated by SR-A. These results question the use of fluorescently labeled bacteria as valid replacements for live bacteria. The low magnitude of opsonin-independent phagocytosis of Ec and unimpaired phagocytosis of Sa in SR-A- or CD36-deficient macrophages indicate that the defect in this process might not be responsible for the reported impaired bacteria clearance in mice deficient in these receptors. We postulate that this impairment might result to a larger extent from inhibition of intracellular bacteria killing caused by pro-inflammatory cytokines, produced in excessive amounts by SR-deficient cells in response to bacterial products.
Collapse
Affiliation(s)
- Angelika Peruń
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Rafał Biedroń
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Maciej K Konopiński
- Institute of Nature Conservation, Polish Academy of Sciences, Cracow, Poland
| | - Anna Białecka
- Centre of Microbiological Research and Autovaccines, Cracow, Poland
| | - Janusz Marcinkiewicz
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Szczepan Józefowski
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
33
|
Dodd CE, Pyle CJ, Glowinski R, Rajaram MVS, Schlesinger LS. CD36-Mediated Uptake of Surfactant Lipids by Human Macrophages Promotes Intracellular Growth of Mycobacterium tuberculosis. THE JOURNAL OF IMMUNOLOGY 2016; 197:4727-4735. [PMID: 27913648 DOI: 10.4049/jimmunol.1600856] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/19/2016] [Indexed: 12/22/2022]
Abstract
Mycobacterium tuberculosis imposes a large global health burden as the airborne agent of tuberculosis. Mycobacterium tuberculosis has been flourishing in human populations for millennia and is therefore highly adapted to the lung environment. Alveolar macrophages, a major host cell niche for M. tuberculosis, are not only phagocytose inhaled microbes and particulate matter but are also crucial in catabolizing lung surfactant, a lipid-protein complex that lines the alveolar spaces. Because macrophage host defense properties can be regulated by surfactant and M. tuberculosis can use host lipids as a carbon source during infection, we sought to determine the receptor(s) involved in surfactant lipid uptake by human macrophages and whether the presence of those lipids within macrophages prior to infection with M. tuberculosis enhances bacterial growth. We show that preformed scavenger receptor CD36 is redistributed to the cell membrane following exposure to surfactant lipids and surfactant protein A. Subsequently, surfactant lipids and/or surfactant protein A enhance CD36 transcript and protein levels. We show that CD36 participates in surfactant lipid uptake by human macrophages, as CD36 knockdown reduces uptake of dipalmitoylphosphatidylcholine, the most prevalent surfactant lipid species. Finally, exposing human macrophages to surfactant lipids prior to infection augments M. tuberculosis growth in a CD36-dependent manner. Thus, we provide evidence that CD36 mediates surfactant lipid uptake by human macrophages and that M. tuberculosis exploits this function for growth.
Collapse
Affiliation(s)
- Claire E Dodd
- Department of Microbiology, The Ohio State University, Columbus, OH 43210; and.,The Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Charlie J Pyle
- The Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Rebecca Glowinski
- The Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Murugesan V S Rajaram
- The Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Larry S Schlesinger
- Department of Microbiology, The Ohio State University, Columbus, OH 43210; and .,The Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
34
|
Cooper GE, Pounce ZC, Wallington JC, Bastidas-Legarda LY, Nicholas B, Chidomere C, Robinson EC, Martin K, Tocheva AS, Christodoulides M, Djukanovic R, Wilkinson TMA, Staples KJ. Viral Inhibition of Bacterial Phagocytosis by Human Macrophages: Redundant Role of CD36. PLoS One 2016; 11:e0163889. [PMID: 27701435 PMCID: PMC5049764 DOI: 10.1371/journal.pone.0163889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/18/2016] [Indexed: 11/18/2022] Open
Abstract
Macrophages are essential to maintaining lung homoeostasis and recent work has demonstrated that influenza-infected lung macrophages downregulate their expression of the scavenger receptor CD36. This receptor has also been shown to be involved in phagocytosis of Streptococcus pneumoniae, a primary agent associated with pneumonia secondary to viral infection. The aim of this study was to investigate the role of CD36 in the effects of viral infection on macrophage phagocytic function. Human monocyte-derived macrophages (MDM) were exposed to H3N2 X31 influenza virus, M37 respiratory syncytial virus (RSV) or UV-irradiated virus. No infection of MDM was seen upon exposure to UV-irradiated virus but incubation with live X31 or M37 resulted in significant levels of viral detection by flow cytometry or RT-PCR respectively. Infection resulted in significantly diminished uptake of S. pneumoniae by MDM and significantly decreased expression of CD36 at both the cell surface and mRNA level. Concurrently, there was a significant increase in IFNβ gene expression in response to infection and we observed a significant decrease in bacterial phagocytosis (p = 0.031) and CD36 gene expression (p = 0.031) by MDM cultured for 24 h in 50IU/ml IFNβ. Knockdown of CD36 by siRNA resulted in decreased phagocytosis, but this was mimicked by transfection reagent alone. When MDM were incubated with CD36 blocking antibodies no effect on phagocytic ability was observed. These data indicate that autologous IFNβ production by virally-infected cells can inhibit bacterial phagocytosis, but that decreased CD36 expression by these cells does not play a major role in this functional deficiency.
Collapse
Affiliation(s)
- Grace E. Cooper
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Zoe C. Pounce
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Joshua C. Wallington
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Leidy Y. Bastidas-Legarda
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Ben Nicholas
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Chiamaka Chidomere
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Emily C. Robinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Kirstin Martin
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Anna S. Tocheva
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Myron Christodoulides
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Ratko Djukanovic
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Karl J. Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Guillou A, Troha K, Wang H, Franc NC, Buchon N. The Drosophila CD36 Homologue croquemort Is Required to Maintain Immune and Gut Homeostasis during Development and Aging. PLoS Pathog 2016; 12:e1005961. [PMID: 27780230 PMCID: PMC5079587 DOI: 10.1371/journal.ppat.1005961] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/29/2016] [Indexed: 12/11/2022] Open
Abstract
Phagocytosis is an ancient mechanism central to both tissue homeostasis and immune defense. Both the identity of the receptors that mediate bacterial phagocytosis and the nature of the interactions between phagocytosis and other defense mechanisms remain elusive. Here, we report that Croquemort (Crq), a Drosophila member of the CD36 family of scavenger receptors, is required for microbial phagocytosis and efficient bacterial clearance. Flies mutant for crq are susceptible to environmental microbes during development and succumb to a variety of microbial infections as adults. Crq acts parallel to the Toll and Imd pathways to eliminate bacteria via phagocytosis. crq mutant flies exhibit enhanced and prolonged immune and cytokine induction accompanied by premature gut dysplasia and decreased lifespan. The chronic state of immune activation in crq mutant flies is further regulated by negative regulators of the Imd pathway. Altogether, our data demonstrate that Crq plays a key role in maintaining immune and organismal homeostasis.
Collapse
Affiliation(s)
- Aurélien Guillou
- Department of Entomology, Cornell University, Ithaca, NY, United States Of America
| | - Katia Troha
- Department of Entomology, Cornell University, Ithaca, NY, United States Of America
| | - Hui Wang
- Department of Cell & Molecular Biology, The Scripps Research Institute, La Jolla, CA, United States Of America
| | - Nathalie C. Franc
- Department of Cell & Molecular Biology, The Scripps Research Institute, La Jolla, CA, United States Of America
| | - Nicolas Buchon
- Department of Entomology, Cornell University, Ithaca, NY, United States Of America
| |
Collapse
|
36
|
Olonisakin TF, Li H, Xiong Z, Kochman EJK, Yu M, Qu Y, Hulver M, Kolls JK, St Croix C, Doi Y, Nguyen MH, Shanks RMQ, Mallampalli RK, Kagan VE, Ray A, Silverstein RL, Ray P, Lee JS. CD36 Provides Host Protection Against Klebsiella pneumoniae Intrapulmonary Infection by Enhancing Lipopolysaccharide Responsiveness and Macrophage Phagocytosis. J Infect Dis 2016; 214:1865-1875. [PMID: 27683817 DOI: 10.1093/infdis/jiw451] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/19/2016] [Indexed: 02/07/2023] Open
Abstract
Klebsiella pneumoniae remains an important cause of intrapulmonary infection and invasive disease worldwide. K. pneumoniae can evade serum killing and phagocytosis primarily through the expression of a polysaccharide capsule, but its pathogenicity is also influenced by host factors. We examined whether CD36, a scavenger receptor that recognizes pathogen and modified self ligands, is a host determinant of K. pneumoniae pathogenicity. Despite differences in serum sensitivity and virulence of 3 distinct K. pneumoniae (hypermucoviscous K1, research K2, and carbapenemase-producing ST258) strains, the absence of CD36 significantly increased host susceptibility to acute intrapulmonary infection by K. pneumoniae, regardless of strain. We demonstrate that CD36 enhances LPS responsiveness to K. pneumoniae to increase downstream cytokine production and macrophage phagocytosis that is independent of polysaccharide capsular antigen. Our study provides new insights into host determinants of K. pneumoniae pathogenicity and raises the possibility that functional mutations in CD36 may predispose individuals to K. pneumoniae syndromes.
Collapse
Affiliation(s)
- Tolani F Olonisakin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Huihua Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Zeyu Xiong
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | - Minting Yu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Yanyan Qu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mei Hulver
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | | | | | | | - Robert M Q Shanks
- Department of Ophthalmology.,Department of Microbiology and Molecular Genetics
| | - Rama K Mallampalli
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Valerian E Kagan
- Department of Environmental Occupational Health, Graduate School of Public Health
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Roy L Silverstein
- Department of Medicine, Medical College of Wisconsin.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee
| | - Prabir Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Janet S Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Vascular Medicine Institute, University of Pittsburgh, Pennsylvania
| |
Collapse
|
37
|
Maier BB, Hladik A, Lakovits K, Korosec A, Martins R, Kral JB, Mesteri I, Strobl B, Müller M, Kalinke U, Merad M, Knapp S. Type I interferon promotes alveolar epithelial type II cell survival during pulmonary Streptococcus pneumoniae infection and sterile lung injury in mice. Eur J Immunol 2016; 46:2175-86. [PMID: 27312374 DOI: 10.1002/eji.201546201] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/17/2016] [Accepted: 06/13/2016] [Indexed: 12/22/2022]
Abstract
Protecting the integrity of the lung epithelial barrier is essential to ensure respiration and proper oxygenation in patients suffering from various types of lung inflammation. Type I interferon (IFN-I) has been associated with pulmonary epithelial barrier function, however, the mechanisms and involved cell types remain unknown. We aimed to investigate the importance of IFN-I with respect to its epithelial barrier strengthening function to better understand immune-modulating effects in the lung with potential medical implications. Using a mouse model of pneumococcal pneumonia, we revealed that IFN-I selectively protects alveolar epithelial type II cells (AECII) from inflammation-induced cell death. Mechanistically, signaling via the IFN-I receptor on AECII is sufficient to promote AECII survival. The net effects of IFN-I are barrier protection, together with diminished tissue damage, inflammation, and bacterial loads. Importantly, we found that the protective role of IFN-I can also apply to sterile acute lung injury, in which loss of IFN-I signaling leads to a significant reduction in barrier function caused by AECII cell death. Our data suggest that IFN-I is an important mediator in lung inflammation that plays a protective role by antagonizing inflammation-associated cell obstruction, thereby strengthening the integrity of the epithelial barrier.
Collapse
Affiliation(s)
- Barbara B Maier
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Laboratory of Infection Biology, Department of Medicine I, Medical University, Vienna, Austria
| | - Anastasiya Hladik
- Laboratory of Infection Biology, Department of Medicine I, Medical University, Vienna, Austria
| | - Karin Lakovits
- Laboratory of Infection Biology, Department of Medicine I, Medical University, Vienna, Austria
| | - Ana Korosec
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Laboratory of Infection Biology, Department of Medicine I, Medical University, Vienna, Austria
| | - Rui Martins
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Laboratory of Infection Biology, Department of Medicine I, Medical University, Vienna, Austria
| | - Julia B Kral
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | | | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, Helmholtz Center for Infection Research, Braunschweig, Germany.,Hannover Medical School, Hannover, Germany
| | - Miriam Merad
- Department of Oncological Science, The Tisch Cancer Institute and the Immunology Institute, Mount Sinai School of Medicine, New York, New York
| | - Sylvia Knapp
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria. .,Laboratory of Infection Biology, Department of Medicine I, Medical University, Vienna, Austria.
| |
Collapse
|
38
|
Liu K, Xu Y, Wang Y, Wei S, Feng D, Huang Q, Zhang S, Liu Z. Developmental expression and immune role of the class B scavenger receptor cd36 in zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 60:91-95. [PMID: 26915754 DOI: 10.1016/j.dci.2016.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 06/05/2023]
Abstract
CD36 is a transmembrane glycoprotein belonging to the scavenger receptor class B family which plays crucial roles in innate immunity. Although CD36 is widely documented in mammals, the study of its functions in fish is still limited. Here we report the identification of a zebrafish cd36 homologue. Zebrafish cd36 has a higher gene expression in the tissues of intestine and liver but very low in kidney and swim bladder. We find cd36 mRNA is maternally expressed and is mainly restricted to the intestine, branchial arches and regions around the lips after the segmentation stage during embryogenesis. Functionally, the recombinant Cd36 corresponding to the large extracellular loop is capable of binding both the Gram-negative and Gram-positive bacteria. These results indicate that zebrafish Cd36 is a microbial-binding molecule. The study expands our knowledge of the function of scavenger receptor molecules in fish innate immune process.
Collapse
Affiliation(s)
- Kai Liu
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Yanping Xu
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Ying Wang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Shulei Wei
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Dong Feng
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Qiaoyan Huang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Shicui Zhang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Zhenhui Liu
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China.
| |
Collapse
|
39
|
CD36 regulates lipopolysaccharide-induced signaling pathways and mediates the internalization of Escherichia coli in cooperation with TLR4 in goat mammary gland epithelial cells. Sci Rep 2016; 6:23132. [PMID: 26976286 PMCID: PMC4791551 DOI: 10.1038/srep23132] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/29/2016] [Indexed: 12/19/2022] Open
Abstract
The scavenger receptor CD36 is involved in pathogen recognition, phagocytosis, and pathogen-induced signaling. This study investigated the relationship between CD36 and TLR4 in modifying lipopolysaccharide (LPS)-induced signaling pathways and mediating Escherichia coli (E. coli) endocytosis in primary goat mammary epithelial cells (pGMECs). The manipulation of CD36 expression significantly influenced TLR4 and nuclear factor kappa B (NF-κB) mRNA expression in pGMECs stimulated with LPS for 12 h. NF-κB and activator protein-1 (AP-1) activity was regulated by the manipulation of CD36 expression in LPS-induced pGMECs. However, CD36-mediated AP-1 activation occurred primarily through c-Jun N-terminal kinase (c-JNK). Adaptor proteins and proinflammatory cytokines were also involved in these signaling pathways and acted by regulating CD36 expression in LPS-stimulated cells. Moreover, CD36 cooperated with TLR4 in TLR4-mediated phagocytosis following E. coli simulation, but this complex was not induced by LPS treatment. Our study is the first to illuminate CD36 as a scavenger receptor in ruminants. Additionally, this study indicates that CD36 plays a vital role in the LPS-induced activation of downstream signaling cascades and mediates E. coli phagocytosis via TLR4 in pGMECs, which offers a novel treatment strategy for mastitis.
Collapse
|
40
|
Castleman MJ, Febbraio M, Hall PR. CD36 Is Essential for Regulation of the Host Innate Response to Staphylococcus aureus α-Toxin-Mediated Dermonecrosis. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26223653 DOI: 10.4049/jimmunol.1500500] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Staphylococcus aureus is the primary cause of skin and skin structure infections (SSSIs) in the United States. α-Hemolysin (Hla), a pore-forming toxin secreted by S. aureus and a major contributor to tissue necrosis, prompts recruitment of neutrophils critical for host defense against S. aureus infections. However, the failure to clear apoptotic neutrophils can result in damage to host tissues, suggesting that mechanisms of neutrophil clearance are essential to limiting Hla-mediated dermonecrosis. We hypothesized that CD36, a scavenger receptor which facilitates recognition of apoptosing cells, would play a significant role in regulating Hla-mediated inflammation and tissue injury during S. aureus SSSI. In this study, we show that CD36 on macrophages negatively regulates dermonecrosis caused by Hla-producing S. aureus. This regulation is independent of bacterial burden, as CD36 also limits dermonecrosis caused by intoxication with sterile bacterial supernatant or purified Hla. Dermonecrotic lesions of supernatant intoxicated CD36(-/-) mice are significantly larger, with increased neutrophil accumulation and IL-1β expression, compared with CD36(+/+) (wild-type) mice. Neutrophil depletion of CD36(-/-) mice prevents this phenotype, demonstrating the contribution of neutrophils to tissue injury in this model. Furthermore, administration of CD36(+/+) but not CD36(-/-) macrophages near the site of intoxication reduces dermonecrosis, IL-1β production and neutrophil accumulation to levels seen in wild-type mice. This therapeutic effect is reversed by inhibiting actin polymerization in the CD36(+/+) macrophages, supporting a mechanism of action whereby CD36-dependent macrophage phagocytosis of apoptotic neutrophils regulates Hla-mediated dermonecrosis. Taken together, these data demonstrate that CD36 is essential for controlling the host innate response to S. aureus skin infection.
Collapse
Affiliation(s)
- Moriah J Castleman
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131; and
| | - Maria Febbraio
- School of Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Pamela R Hall
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131; and
| |
Collapse
|
41
|
Soriano-Romaní L, Contreras-Ruiz L, García-Posadas L, López-García A, Masli S, Diebold Y. Inflammatory Cytokine-Mediated Regulation of Thrombospondin-1 and CD36 in Conjunctival Cells. J Ocul Pharmacol Ther 2015; 31:419-28. [PMID: 26154920 DOI: 10.1089/jop.2015.0029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Increased expression of transforming growth factor-β2 (TGF-β2) is reported in the conjunctiva of dry eye patients with no increase of anti-inflammatory activity of TGF-β2. Our aim was to compare the expression of molecules involved in TGF-β2 activation, thrombospondin-1 (TSP-1) and CD36, during murine and human conjunctival inflammation. METHODS Human conjunctival tissue from cadaveric donors, human conjunctival epithelial primary cells and fibroblasts, and murine conjunctivas were immunostained for TSP-1, CD36, or TGF-β2. Inflamed conjunctival tissues were obtained from C57BL/6 wild-type (WT) mice induced to develop experimental dry eye (EDE) with 10 days of desiccating conditions and scopolamine injections and TSP-1-deficient (TSP1(-/-)) mice, which spontaneously develop Sjögren's syndrome-associated conjunctival inflammation with age. Immunostaining intensities were compared using ImageJ software. Cultures of human conjunctival fibroblasts were stimulated with IL-1β and both secreted protein and message levels of TSP-1, CD36, and TGF-β2 were analyzed. RESULTS TSP-1 and CD36 were detectable in human and murine conjunctival tissues as well as primary conjunctival epithelial cells and fibroblasts. Increased conjunctival immunostaining of TGF-β2 and reduced CD36 were detected in EDE mice compared with WT mice. Interestingly, increased TGF-β2 and CD36 conjunctival immunostaining was detected in TSP1(-/-) mice. The expression of TSP-1 and CD36 was downregulated in IL-1β-stimulated conjunctival fibroblasts at both the protein and message level, while active TGF-β2 was undetected. CONCLUSIONS The absence or reduced expression of either of the molecules involved in TGF-β2 activation supports proinflammatory conditions in the conjunctiva. Changes in TSP-1 and CD36 may serve as potential biomarkers of conjunctival inflammation.
Collapse
Affiliation(s)
- Laura Soriano-Romaní
- 1 Ocular Surface Group-IOBA, University of Valladolid , Valladolid, Spain .,2 Biomedical Research Networking Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Laura Contreras-Ruiz
- 3 Department of Ophthalmology, Boston University School of Medicine , Boston, Massachusetts
| | - Laura García-Posadas
- 1 Ocular Surface Group-IOBA, University of Valladolid , Valladolid, Spain .,2 Biomedical Research Networking Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Antonio López-García
- 1 Ocular Surface Group-IOBA, University of Valladolid , Valladolid, Spain .,2 Biomedical Research Networking Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Sharmila Masli
- 3 Department of Ophthalmology, Boston University School of Medicine , Boston, Massachusetts
| | - Yolanda Diebold
- 1 Ocular Surface Group-IOBA, University of Valladolid , Valladolid, Spain .,2 Biomedical Research Networking Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN), Spain
| |
Collapse
|
42
|
Bi WJ, Li DX, Xu YH, Xu S, Li J, Zhao XF, Wang JX. Scavenger receptor B protects shrimp from bacteria by enhancing phagocytosis and regulating expression of antimicrobial peptides. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 51:10-21. [PMID: 25697802 DOI: 10.1016/j.dci.2015.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 06/04/2023]
Abstract
Scavenger receptors (SRs) are involved in innate immunity through recognizing pathogen-associated molecular patterns (PAMPs) and in pathogenesis of diseases through interactions with damage-associated molecular patterns (DAMPs). The roles of SRs in invertebrate innate immunity still need to be elucidated. Here we identify a class B scavenger receptor from kuruma shrimp, Marsupenaeus japonicus, designated MjSR-B1. The recombinant MjSR-B1 agglutinated bacteria in a calcium dependent manner and bound lipopolysaccharide and lipoteichoic acid. After knockdown of MjSR-B1, both the bacterial clearance and phagocytotic ability of M. japonicus against V. anguillarum and S. aureus were impaired, and several phagocytosis related genes were downregulated. The expression levels of antimicrobial peptides were also downregulated. Overexpression of MjSR-B1 led to enhanced bacterial clearance, phagocytosis rate and upregulation of phagocytosis-related and antimicrobial peptide genes. However, overexpression of mutant MjSR-B1ΔC, which lacks the carboxyl tail of MjSR-B1, had none of these effects. Our results indicate that MjSR-B1 can protect shrimp from bacteria by promoting phagocytosis and by enhancing the expression of antimicrobial peptides.
Collapse
Affiliation(s)
- Wen-Jie Bi
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Dian-Xiang Li
- Biotechnology Department, School of Biological Sciences and Biotechnology, University of Jinan, Jinan 250022, China
| | - Yi-Hui Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Sen Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Jing Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China.
| |
Collapse
|
43
|
Sanfilippo AM, Furuya Y, Roberts S, Salmon SL, Metzger DW. Allergic Lung Inflammation Reduces Tissue Invasion and Enhances Survival from Pulmonary Pneumococcal Infection in Mice, Which Correlates with Increased Expression of Transforming Growth Factor β1 and SiglecF(low) Alveolar Macrophages. Infect Immun 2015; 83:2976-83. [PMID: 25964474 PMCID: PMC4468552 DOI: 10.1128/iai.00142-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/04/2015] [Indexed: 11/20/2022] Open
Abstract
Asthma is generally thought to confer an increased risk for invasive pneumococcal disease (IPD) in humans. However, recent reports suggest that mortality rates from IPD are unaffected in patients with asthma and that chronic obstructive pulmonary disease (COPD), a condition similar to asthma, protects against the development of complicated pneumonia. To clarify the effects of asthma on the subsequent susceptibility to pneumococcal infection, ovalbumin (OVA)-induced allergic lung inflammation (ALI) was induced in mice followed by intranasal infection with A66.1 serotype 3 Streptococcus pneumoniae. Surprisingly, mice with ALI were significantly more resistant to lethal infection than non-ALI mice. The heightened resistance observed following ALI correlated with enhanced early clearance of pneumococci from the lung, decreased bacterial invasion from the airway into the lung tissue, a blunted inflammatory cytokine and neutrophil response to infection, and enhanced expression of transforming growth factor β1 (TGF-β1). Neutrophil depletion prior to infection had no effect on enhanced early bacterial clearance or resistance to IPD in mice with ALI. Although eosinophils recruited into the lung during ALI appeared to be capable of phagocytizing bacteria, neutralization of interleukin-5 (IL-5) to inhibit eosinophil recruitment likewise had no effect on early clearance or survival following infection. However, enhanced resistance was associated with an increase in levels of clodronate-sensitive, phagocytic SiglecF(low) alveolar macrophages within the airways following ALI. These findings suggest that, while the risk of developing IPD may actually be decreased in patients with acute asthma, additional clinical data are needed to better understand the risk of IPD in patients with different asthma phenotypes.
Collapse
Affiliation(s)
- Alan M Sanfilippo
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Yoichi Furuya
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sean Roberts
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sharon L Salmon
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Dennis W Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
44
|
Soriano-Romaní L, García-Posadas L, López-García A, Paraoan L, Diebold Y. Thrombospondin-1 induces differential response in human corneal and conjunctival epithelial cells lines under in vitro inflammatory and apoptotic conditions. Exp Eye Res 2015; 134:1-14. [PMID: 25753839 DOI: 10.1016/j.exer.2015.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/06/2015] [Accepted: 03/05/2015] [Indexed: 11/19/2022]
Abstract
Recently, thrombospondin-1 (TSP-1) has been reported to be critical for maintaining a healthy ocular surface. The purpose of the study was to characterize the expression of TSP-1 and of its receptors CD36 and CD47 in corneal and conjunctival epithelial cells and determine the effect of exogenous TSP-1 treatment on these cells, following the induction of inflammation- and apoptosis-related changes. The expression of TSP-1, CD36 and CD47 by corneal and conjunctival cell lines was firstly characterized by ELISA, immunofluorescence analysis, Western blotting and reverse transcription polymerase chain reaction (RT-PCR). Benzalkonium chloride (BAC) exposure for 5 or 15 min was used as pro-inflammatory and pro-apoptotic stimulus for corneal or conjunctival epithelial cells, respectively. To analyze inflammation and apoptosis-related changes, IL-6 and TGF-β2 secretion determined by ELISA was used as inflammatory markers, while activated caspase-3/7 levels and cell viability, determined by CellEvent™ Caspase-3/7 Green Detection Reagent and XTT cytotoxicity assay, respectively, were used as apoptotic markers. Changes in CD36 and CD47 mRNA expression were quantified by real time RT-PCR. Corneal epithelial cells secreted and expressed higher protein levels of TSP-1 than conjunctival epithelial cells, although TSP-1 mRNA expression levels were similar and had lower CD36 and CD47, both at protein and mRNA levels. Both cell lines responded to exogenous TSP-1 treatment increasing CD36 at protein and mRNA levels. Blocking experiments revealed a predominance of TSP-1/CD47 rather than TSP-1/CD36 interactions to up-regulate CD36 levels in conjunctival epithelial cells, but not in corneal epithelial cells. BAC exposure increased IL-6 secretion and caspase-3/7 levels and decreased cell viability in both, corneal and conjunctival epithelial cells. Moreover, BAC exposure increased latent TGF-β2 levels in conjunctival epithelial cells. Interestingly, CD36 mRNA expression was down-regulated after BAC exposure in both cell lines. Exogenous TSP-1 treatment reduced TGF-β2 up-regulated levels by BAC exposure in conjunctival epithelial cells and less pronounced reduced IL-6 in BAC-exposed corneal epithelial cells. The effect on CD36 and CD47 regulation was less pronounced or even opposite depending on the inflammation- and apoptosis-related markers tested. Our results show evidence of the capacity of corneal and conjunctival epithelial cells to respond to TSP-1 via CD36 or CD47. Experimental simulation of inflammation- and apoptosis-related conditions changed the effects differentially elicited by TSP-1 on corneal and conjunctival epithelial cells, suggesting an unexpected and relevant contribution of TSP-1 on ocular surface homeostasis regulation.
Collapse
Affiliation(s)
| | | | | | - Luminita Paraoan
- Department of Eye and Vision Science, University of Liverpool, Liverpool, UK
| | - Yolanda Diebold
- Ocular Surface Group-IOBA, University of Valladolid, Valladolid, Spain.
| |
Collapse
|
45
|
Gawish R, Martins R, Böhm B, Wimberger T, Sharif O, Lakovits K, Schmidt M, Knapp S. Triggering receptor expressed on myeloid cells-2 fine-tunes inflammatory responses in murine Gram-negative sepsis. FASEB J 2014; 29:1247-57. [PMID: 25477281 DOI: 10.1096/fj.14-260067] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 11/13/2014] [Indexed: 12/18/2022]
Abstract
During infections, TLR-mediated responses require tight regulation to allow for pathogen removal, while preventing overwhelming inflammation and immunopathology. The triggering receptor expressed on myeloid cells (TREM)-2 negatively regulates inflammation by macrophages and impacts on phagocytosis, but the function of endogenous TREM-2 during infections is poorly understood. We investigated TREM-2's role in regulating TLR4-mediated inflammation by studying wild-type and TREM-2(-/-) mice challenged with LPS and found TREM-2 to dampen early inflammation. Augmented early inflammation in TREM-2(-/-) animals was followed by an accelerated resolution and ultimately improved survival, associated with the induction of the negative regulator A20. Upon infection with Escherichia coli, the otherwise beneficial effect of an exaggerated early immune response in TREM-2(-/-) animals was counteracted by a 50% reduction in bacterial phagocytosis. In line with this, TREM-2(-/-) peritoneal macrophages (PMs) exhibited augmented inflammation following TLR4 stimulation, demonstrating the presence and negative regulatory functionality of TREM-2 on primary PMs. Significantly, we identified a high turnover rate because TREM-2 RNA is 25-fold down-regulated and the protein proteasomally degraded upon LPS encounter, thus ensuring a tightly regulated and versatile system that modulates inflammation. Our results illustrate TREM-2's effects on infection-triggered inflammation and identify TREM-2 as a potential target to prevent overwhelming inflammation while preserving antibacterial-effector functions.
Collapse
Affiliation(s)
- Riem Gawish
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Rui Martins
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Benedikta Böhm
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Terje Wimberger
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Omar Sharif
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Karin Lakovits
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Mariane Schmidt
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| | - Sylvia Knapp
- *Ce-M-M-, Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria; and National Food Institute, Division for Epidemiology and Microbial Genomics, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
46
|
Vassal-Stermann E, Lacroix M, Gout E, Laffly E, Pedersen CM, Martin L, Amoroso A, Schmidt RR, Zähringer U, Gaboriaud C, Di Guilmi AM, Thielens NM. Human L-ficolin recognizes phosphocholine moieties of pneumococcal teichoic acid. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:5699-708. [PMID: 25344472 DOI: 10.4049/jimmunol.1400127] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Human L-ficolin is a soluble protein of the innate immune system able to sense pathogens through its fibrinogen (FBG) recognition domains and to trigger activation of the lectin complement pathway through associated serine proteases. L-Ficolin has been previously shown to recognize pneumococcal clinical isolates, but its ligands and especially its molecular specificity remain to be identified. Using solid-phase binding assays, serum and recombinant L-ficolins were shown to interact with serotype 2 pneumococcal strain D39 and its unencapsulated R6 derivative. Incubation of both strains with serum triggered complement activation, as measured by C4b and C3b deposition, which was decreased by using ficolin-depleted serum. Recombinant L-ficolin and its FBG-like recognition domain bound to isolated pneumococcal cell wall extracts, whereas binding to cell walls depleted of teichoic acid (TA) was decreased. Both proteins were also shown to interact with two synthetic TA compounds, each comprising part structures of the complete lipoteichoic acid molecule with two PCho residues. Competition studies and direct interaction measurements by surface plasmon resonance identified PCho as a novel L-ficolin ligand. Structural analysis of complexes of the FBG domain of L-ficolin and PCho revealed that the phosphate moiety interacts with amino acids previously shown to define an acetyl binding site. Consequently, binding of L-ficolin to immobilized acetylated BSA was inhibited by PCho and synthetic TA. Binding of serum L-ficolin to immobilized synthetic TA and PCho-conjugated BSA triggered activation of the lectin complement pathway, thus further supporting the hypothesis of L-ficolin involvement in host antipneumococcal defense.
Collapse
Affiliation(s)
- Emilie Vassal-Stermann
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Monique Lacroix
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Evelyne Gout
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Emmanuelle Laffly
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | | | - Lydie Martin
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Ana Amoroso
- Centre for Protein Engineering, Department of Life Sciences, University of Liege, B4000 Liege, Belgium
| | - Richard R Schmidt
- Department of Chemistry, University of Konstanz, D-78457 Konstanz, Germany; Chemistry Department, King Abdulaziz University of Jeddah, 21589 Jeddah, Saudi Arabia; and
| | - Ulrich Zähringer
- Division of Immunochemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, D-23845 Borstel, Germany
| | - Christine Gaboriaud
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Anne-Marie Di Guilmi
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France;
| | - Nicole M Thielens
- University of Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France; Centre National de la Recherche Scientifique, Institut de Biologie Structurale, F-38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| |
Collapse
|
47
|
Benard EL, Roobol SJ, Spaink HP, Meijer AH. Phagocytosis of mycobacteria by zebrafish macrophages is dependent on the scavenger receptor Marco, a key control factor of pro-inflammatory signalling. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:223-233. [PMID: 25086293 DOI: 10.1016/j.dci.2014.07.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 06/03/2023]
Abstract
Scavenger receptors on the cell surface of macrophages play an important role in host defence through their ability to bind microbial ligands and induce phagocytosis. Concurrently, signal transduction pathways are initiated that aid in defence mechanisms against the invading microbe. Here we report on the function of scavenger receptor Marco (Macrophage receptor with collagenous structure) during infection of zebrafish embryos with Mycobacterium marinum, a close relative of M. tuberculosis. Morpholino knockdown demonstrates that Marco is required for the rapid phagocytosis of M. marinum following intravenous infection. Furthermore, gene expression analysis shows that Marco controls the initial transient pro-inflammatory response to M. marinum and remains a determining factor for the immune response signature at later stages of infection. Increased bacterial burden following marco knockdown indicates that this scavenger receptor is important for control of M. marinum growth, likely due to delayed phagocytosis and reduced pro-inflammatory signalling observed under conditions of Marco deficiency.
Collapse
Affiliation(s)
- Erica L Benard
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Stefan J Roobol
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Herman P Spaink
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Annemarie H Meijer
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
48
|
Cole J, Aberdein J, Jubrail J, Dockrell DH. The role of macrophages in the innate immune response to Streptococcus pneumoniae and Staphylococcus aureus: mechanisms and contrasts. Adv Microb Physiol 2014; 65:125-202. [PMID: 25476766 DOI: 10.1016/bs.ampbs.2014.08.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophages are critical mediators of innate immune responses against bacteria. The Gram-positive bacteria Streptococcus pneumoniae and Staphylococcus aureus express a range of virulence factors, which challenge macrophages' immune competence. We review how macrophages respond to this challenge. Macrophages employ a range of strategies to phagocytose and kill each pathogen. When the macrophages capacity to clear bacteria is overwhelmed macrophages play important roles in orchestrating the inflammatory response through pattern recognition receptor-mediated responses. Macrophages also ensure the inflammatory response is tightly constrained, to avoid tissue damage, and play an important role in downregulating the inflammatory response once initial bacterial replication is controlled.
Collapse
Affiliation(s)
- Joby Cole
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Jody Aberdein
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Jamil Jubrail
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - David H Dockrell
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom.
| |
Collapse
|
49
|
Sharif O, Gawish R, Warszawska JM, Martins R, Lakovits K, Hladik A, Doninger B, Brunner J, Korosec A, Schwarzenbacher RE, Berg T, Kralovics R, Colinge J, Mesteri I, Gilfillan S, Salmaggi A, Verschoor A, Colonna M, Knapp S. The triggering receptor expressed on myeloid cells 2 inhibits complement component 1q effector mechanisms and exerts detrimental effects during pneumococcal pneumonia. PLoS Pathog 2014; 10:e1004167. [PMID: 24945405 PMCID: PMC4055749 DOI: 10.1371/journal.ppat.1004167] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 04/07/2014] [Indexed: 11/18/2022] Open
Abstract
Phagocytosis and inflammation within the lungs is crucial for host defense during bacterial pneumonia. Triggering receptor expressed on myeloid cells (TREM)-2 was proposed to negatively regulate TLR-mediated responses and enhance phagocytosis by macrophages, but the role of TREM-2 in respiratory tract infections is unknown. Here, we established the presence of TREM-2 on alveolar macrophages (AM) and explored the function of TREM-2 in the innate immune response to pneumococcal infection in vivo. Unexpectedly, we found Trem-2(-/-) AM to display augmented bacterial phagocytosis in vitro and in vivo compared to WT AM. Mechanistically, we detected that in the absence of TREM-2, pulmonary macrophages selectively produced elevated complement component 1q (C1q) levels. We found that these increased C1q levels depended on peroxisome proliferator-activated receptor-δ (PPAR-δ) activity and were responsible for the enhanced phagocytosis of bacteria. Upon infection with S. pneumoniae, Trem-2(-/-) mice exhibited an augmented bacterial clearance from lungs, decreased bacteremia and improved survival compared to their WT counterparts. This work is the first to disclose a role for TREM-2 in clinically relevant respiratory tract infections and demonstrates a previously unknown link between TREM-2 and opsonin production within the lungs.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Line, Transformed
- Cells, Cultured
- Complement C1q/genetics
- Complement C1q/metabolism
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Lung/cytology
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/pathology
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Infiltration
- PPAR gamma/metabolism
- Phagocytosis
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/metabolism
- Pneumonia, Pneumococcal/pathology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Respiratory Mucosa/cytology
- Respiratory Mucosa/immunology
- Respiratory Mucosa/metabolism
- Respiratory Mucosa/pathology
- Survival Analysis
Collapse
Affiliation(s)
- Omar Sharif
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- * E-mail: (OS); (SK)
| | - Riem Gawish
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Joanna M. Warszawska
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Rui Martins
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Karin Lakovits
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Bianca Doninger
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Julia Brunner
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Ana Korosec
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Roland E. Schwarzenbacher
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Tiina Berg
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Robert Kralovics
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jacques Colinge
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ildiko Mesteri
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrea Salmaggi
- Department of Clinical Neurosciences, Istituto Nazionale Neurologico Carlo Besta, Milano, Italy
| | - Admar Verschoor
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sylvia Knapp
- CeMM - Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- * E-mail: (OS); (SK)
| |
Collapse
|
50
|
Abstract
As investigations into the innate immune responses that lead to allergic sensitization become better defined, there is a need to determine how allergens could interact with pattern recognition receptors that bind non-proteinaceous moieties. Many important allergens are not covalently bound to lipid or carbohydrate, but have structures belonging to lipid, glycan and glycolipid-binding families. These include ML-domain proteins, lipopolysaccharide-binding/cell permeability-increasing proteins, von Ebner gland lipocalins, salivary lipocalins/major urinary proteins, plant pathogenesis-related proteins PR-5 and -10, uteroglobins, non-specific lipid transfer proteins, large lipid transfer proteins and proteins with chitin and other carbohydrate-binding modules. The binding expected is overviewed with regard to importance of the allergens and their ability to elicit responses proposed from experimental models. The evidence compiled showing that allergens from the same source sensitize for different types of adaptive immune responses supports the concept that individual allergens within these sources have their own distinctive interactions with innate immunity.
Collapse
|